Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Ann Coloproctol ; 40(2): 121-135, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38712438

ABSTRACT

PURPOSE: Colorectal cancer (CRC) is the most frequent cancer with limited therapeutic achievements. Recently, adoptive cellular immunotherapy has been developed as an antitumor therapy. However, its efficacy has not been tested in CRC. This study investigated the ability of an immune cell cocktail of dendritic cells (DCs), T cells, and natural killer (NK) cells to overcome immunological hurdles and improve the therapeutic efficacy of cell therapy for CRC. METHODS: CRC lysate-pulsed monocyte-derived DCs (Mo-DCs), CRC antigen-specifically expanded T cells (CTL), and in vitro-expanded NK cells were cultured from patient peripheral blood mononuclear cells (PBMC). The ability of the combined immune cells to kill autologous tumor cells was investigated by co-culturing the combined immune cells with patient-derived tumor cells. RESULTS: The Mo-DCs produced expressed T cell co-stimulating molecules like CD80, CD86, human leukocyte antigen (HLA)-DR and HLA-ABC, at high levels and were capable of activating naive T cells. The expanded T cells were predominantly CD8 T cells with high levels of CD8 effector memory cells and low levels of regulatory T cells. The NK cells expressed high levels of activating receptors and were capable of killing other cancer cell lines (K562 and HT29). The immune cell cocktail demonstrated a higher ability to kill autologous tumor cells than single types. An in vivo preclinical study confirmed the safety of the combined immune cell adaptive therapy showing no therapy-related death or general toxicity symptoms. CONCLUSION: The results suggested that combined immune cell adaptive therapy could overcome the limited efficacy of cell immunotherapy.

2.
Nat Commun ; 14(1): 6493, 2023 10 14.
Article in English | MEDLINE | ID: mdl-37838725

ABSTRACT

Misfolded protein aggregates may cause toxic proteinopathy, including autosomal dominant tubulointerstitial kidney disease due to uromodulin mutations (ADTKD-UMOD), a leading hereditary kidney disease. There are no targeted therapies. In our generated mouse model recapitulating human ADTKD-UMOD carrying a leading UMOD mutation, we show that autophagy/mitophagy and mitochondrial biogenesis are impaired, leading to cGAS-STING activation and tubular injury. Moreover, we demonstrate that inducible tubular overexpression of mesencephalic astrocyte-derived neurotrophic factor (MANF), a secreted endoplasmic reticulum protein, after the onset of disease stimulates autophagy/mitophagy, clears mutant UMOD, and promotes mitochondrial biogenesis through p-AMPK enhancement, thus protecting kidney function in our ADTKD mouse model. Conversely, genetic ablation of MANF in the mutant thick ascending limb tubular cells worsens autophagy suppression and kidney fibrosis. Together, we have discovered MANF as a biotherapeutic protein and elucidated previously unknown mechanisms of MANF in the regulation of organelle homeostasis, which may have broad therapeutic applications to treat various proteinopathies.


Subject(s)
Polycystic Kidney Diseases , Humans , Mice , Animals , Autophagy/genetics , Homeostasis , Fibrosis , Nerve Growth Factors/genetics
3.
bioRxiv ; 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36711449

ABSTRACT

Misfolded protein aggregates may cause toxic proteinopathy, including autosomal dominant tubulointerstitial kidney disease due to uromodulin mutations (ADTKD- UMOD ), one of the leading hereditary kidney diseases, and Alzheimer’s disease etc. There are no targeted therapies. ADTKD is also a genetic form of renal fibrosis and chronic kidney disease, which affects 500 million people worldwide. For the first time, in our newly generated mouse model recapitulating human ADTKD- UMOD carrying a leading UMOD deletion mutation, we show that autophagy/mitophagy and mitochondrial biogenesis are severely impaired, leading to cGAS- STING activation and tubular injury. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a novel endoplasmic reticulum stress-regulated secreted protein. We provide the first study that inducible tubular overexpression of MANF after the onset of disease stimulates autophagy/mitophagy and clearance of the misfolded UMOD, and promotes mitochondrial biogenesis through p-AMPK enhancement, resulting in protection of kidney function. Conversely, genetic ablation of endogenous MANF upregulated in the mutant mouse and human tubular cells worsens autophagy suppression and kidney fibrosis. Together, we discover MANF as a novel biotherapeutic protein and elucidate previously unknown mechanisms of MANF in regulating organelle homeostasis to treat ADTKD, which may have broad therapeutic application to treat various proteinopathies.

4.
Proc Natl Acad Sci U S A ; 119(35): e2116505119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994650

ABSTRACT

Albuminuria is a hallmark of glomerular disease of various etiologies. It is not only a symptom of glomerular disease but also a cause leading to glomerulosclerosis, interstitial fibrosis, and eventually, a decline in kidney function. The molecular mechanism underlying albuminuria-induced kidney injury remains poorly defined. In our genetic model of nephrotic syndrome (NS), we have identified CHOP (C/EBP homologous protein)-TXNIP (thioredoxin-interacting protein) as critical molecular linkers between albuminuria-induced ER dysfunction and mitochondria dyshomeostasis. TXNIP is a ubiquitously expressed redox protein that binds to and inhibits antioxidant enzyme, cytosolic thioredoxin 1 (Trx1), and mitochondrial Trx2. However, very little is known about the regulation and function of TXNIP in NS. By utilizing Chop-/- and Txnip-/- mice as well as 68Ga-Galuminox, our molecular imaging probe for detection of mitochondrial reactive oxygen species (ROS) in vivo, we demonstrate that CHOP up-regulation induced by albuminuria drives TXNIP shuttling from nucleus to mitochondria, where it is required for the induction of mitochondrial ROS. The increased ROS accumulation in mitochondria oxidizes Trx2, thus liberating TXNIP to associate with mitochondrial nod-like receptor protein 3 (NLRP3) to activate inflammasome, as well as releasing mitochondrial apoptosis signal-regulating kinase 1 (ASK1) to induce mitochondria-dependent apoptosis. Importantly, inhibition of TXNIP translocation and mitochondrial ROS overproduction by CHOP deletion suppresses NLRP3 inflammasome activation and p-ASK1-dependent mitochondria apoptosis in NS. Thus, targeting TXNIP represents a promising therapeutic strategy for the treatment of NS.


Subject(s)
Albuminuria , Carrier Proteins , Kidney , Mitochondria , Nephrotic Syndrome , Thioredoxins , Transcription Factor CHOP , Albuminuria/complications , Albuminuria/genetics , Albuminuria/prevention & control , Animals , Apoptosis , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Gene Deletion , Inflammasomes/metabolism , Kidney/metabolism , Kidney/pathology , MAP Kinase Kinase Kinase 5/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrotic Syndrome/complications , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Nephrotic Syndrome/prevention & control , Reactive Oxygen Species/metabolism , Thioredoxins/metabolism , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
5.
Am J Physiol Renal Physiol ; 321(2): F236-F244, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34251273

ABSTRACT

Autosomal dominant tubulointerstitial kidney disease (ADTKD)-uromodulin (UMOD) is the most common nonpolycystic genetic kidney disease, but it remains unrecognized due to its clinical heterogeneity and lack of screening test. Moreover, the fact that the clinical feature is a poor predictor of disease outcome further highlights the need for the development of mechanistic biomarkers in ADTKD. However, low abundant urinary proteins secreted by thick ascending limb cells, where UMOD is synthesized, have posed a challenge for the detection of biomarkers in ADTKD-UMOD. In the CRISPR/Cas9-generated murine model and patients with ADTKD-UMOD, we found that immunoglobulin heavy chain-binding protein (BiP), an endoplasmic reticulum chaperone, was exclusively upregulated by mutant UMOD in the thick ascending limb and easily detected by Western blot analysis in the urine at an early stage of disease. However, even the most sensitive ELISA failed to detect urinary BiP in affected individuals. We therefore developed an ultrasensitive, plasmon-enhanced fluorescence-linked immunosorbent assay (p-FLISA) to quantify urinary BiP concentration by harnessing the newly invented ultrabright fluorescent nanoconstruct, termed "plasmonic Fluor." p-FLISA demonstrated that urinary BiP excretion was significantly elevated in patients with ADTKD-UMOD compared with unaffected controls, which may have potential utility in risk stratification, disease activity monitoring, disease progression prediction, and guidance of endoplasmic reticulum-targeted therapies in ADTKD.NEW & NOTEWORTHY Autosomal dominant tubulointerstitial kidney disease (ADTKD)-uromodulin (UMOD) is an underdiagnosed cause of chronic kidney disease (CKD). Lack of ultrasensitive bioanalytical tools has hindered the discovery of low abundant urinary biomarkers in ADTKD. Here, we developed an ultrasensitive plasmon-enhanced fluorescence-linked immunosorbent assay (p-FLISA). p-FLISA demonstrated that secreted immunoglobulin heavy chain-binding protein is an early urinary endoplasmic reticulum stress biomarker in ADTKD-UMOD, which will be valuable in monitoring disease progression and the treatment response in ADTKD.


Subject(s)
Biomarkers/urine , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/urine , Immunosorbent Techniques , Nephritis, Interstitial/urine , Animals , Endoplasmic Reticulum Chaperone BiP , Humans , Mice , Nephritis, Interstitial/genetics , Uromodulin/genetics
6.
J Microbiol Biotechnol ; 30(6): 937-945, 2020 Jun 28.
Article in English | MEDLINE | ID: mdl-32160697

ABSTRACT

N-acyl-homoserine lactone (AHL)-mediated quorum sensing (QS) plays a major role in development of biofilms, which contribute to rise in infections and biofouling in water-related industries. Interference in QS, called quorum quenching (QQ), has recieved a lot of attention in recent years. Rhodococcus spp. are known to have prominent quorum quenching activity and in previous reports it was suggested that this genus possesses multiple QQ enzymes, but only one gene, qsdA, which encodes an AHL-lactonase belonging to phosphotriesterase family, has been identified. Therefore, we conducted a whole genome sequencing and analysis of Rhodococcus sp. BH4 isolated from a wastewater treatment plant. The sequencing revealed another gene encoding a QQ enzyme (named jydB) that exhibited a high AHL degrading activity. This QQ enzyme had a 46% amino acid sequence similarity with the AHL-lactonase (AidH) of Ochrobactrum sp. T63. HPLC analysis and AHL restoration experiments by acidification revealed that the jydB gene encodes an AHL-lactonase which shares the known characteristics of the α/ß hydrolase family. Purified recombinant JydB demonstrated a high hydrolytic activity against various AHLs. Kinetic analysis of JydB revealed a high catalytic efficiency (kcat/KM) against C4-HSL and 3-oxo-C6 HSL, ranging from 1.88 × 106 to 1.45 × 106 M-1 s-1, with distinctly low KM values (0.16 - 0.24 mM). This study affirms that the AHL degrading activity and biofilm inhibition ability of Rhodococcus sp. BH4 may be due to the presence of multiple quorum quenching enzymes, including two types of AHL-lactonases, in addition to AHL-acylase and oxidoreductase, for which the genes have yet to be described.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Quorum Sensing , Rhodococcus/enzymology , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/metabolism , Biofilms/growth & development , Genes, Bacterial , Kinetics , Rhodococcus/genetics , Wastewater/microbiology , Whole Genome Sequencing
7.
Proc Natl Acad Sci U S A ; 116(28): 14154-14163, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31235574

ABSTRACT

Emerging evidence has established primary nephrotic syndrome (NS), including focal segmental glomerulosclerosis (FSGS), as a primary podocytopathy. Despite the underlying importance of podocyte endoplasmic reticulum (ER) stress in the pathogenesis of NS, no treatment currently targets the podocyte ER. In our monogenic podocyte ER stress-induced NS/FSGS mouse model, the podocyte type 2 ryanodine receptor (RyR2)/calcium release channel on the ER was phosphorylated, resulting in ER calcium leak and cytosolic calcium elevation. The altered intracellular calcium homeostasis led to activation of calcium-dependent cytosolic protease calpain 2 and cleavage of its important downstream substrates, including the apoptotic molecule procaspase 12 and podocyte cytoskeletal protein talin 1. Importantly, a chemical compound, K201, can block RyR2-Ser2808 phosphorylation-mediated ER calcium depletion and podocyte injury in ER-stressed podocytes, as well as inhibit albuminuria in our NS model. In addition, we discovered that mesencephalic astrocyte-derived neurotrophic factor (MANF) can revert defective RyR2-induced ER calcium leak, a bioactivity for this ER stress-responsive protein. Thus, podocyte RyR2 remodeling contributes to ER stress-induced podocyte injury. K201 and MANF could be promising therapies for the treatment of podocyte ER stress-induced NS/FSGS.


Subject(s)
Calcium/metabolism , Nephrotic Syndrome/genetics , Nerve Growth Factors/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Albuminuria/drug therapy , Albuminuria/genetics , Albuminuria/pathology , Animals , Calcium Signaling/genetics , Calpain/genetics , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/genetics , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Humans , Mice , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Podocytes/metabolism , Podocytes/pathology , Talin/genetics , Thiazepines/pharmacology
8.
Pediatr Nephrol ; 34(9): 1493-1500, 2019 09.
Article in English | MEDLINE | ID: mdl-30099615

ABSTRACT

The advent of next-generation sequencing (NGS) in recent years has led to a rapid discovery of novel or rare genetic variants in human kidney cell genes, which is transforming the risk assessment, diagnosis, and treatment of kidney disease. Mutations may lead to protein misfolding, disruption of protein trafficking, and endoplasmic reticulum (ER) retention. An imbalance between the load of misfolded proteins and the folding capacity of the ER causes ER stress and unfolded protein response. Mutations in nephrin (NPHS1), podocin (NPHS2), laminin ß2 (LAMB2), and α-actinin-4 (ACTN4) have been shown to induce ER stress in HEK293 cells and podocytes in hereditary nephrotic syndromes; various founder mutations in collagen IV α chains (COL4A) have been demonstrated to activate podocyte ER stress in collagen IV nephropathies; and mutations in uromodulin (UMOD) have been reported to trigger tubular ER stress in autosomal dominant tubulointerstitial kidney disease. Meanwhile, ER resident protein SEC63 may modify disease severity in autosomal dominant polycystic kidney disease. These findings underscore the importance of ER stress in the pathogenesis of monogenic kidney disease. Recently, we have identified mesencephalic astrocyte-derived neurotrophic factor (MANF) and cysteine-rich with EGF-like domains 2 (CRELD2) as urinary ER stress biomarkers in ER stress-mediated kidney diseases.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Kidney Diseases/drug therapy , Nephrology/methods , Precision Medicine/methods , Biomarkers/analysis , DNA Mutational Analysis , Endoplasmic Reticulum Stress/genetics , High-Throughput Nucleotide Sequencing , Humans , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Kidney Diseases/pathology , Mutation , Podocytes/drug effects , Podocytes/pathology
9.
Environ Sci Technol ; 52(11): 6237-6245, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29714471

ABSTRACT

Bacterial quorum quenching (QQ) by means of degrading signaling molecules has been applied to antibiofouling strategies in a membrane bioreactor (MBR) for wastewater treatment. However, the target signaling molecules have been limited to N-acyl homoserine lactones participating in intraspecies quorum sensing. Here, an approach to disrupting autoinducer-2 (AI-2) signaling molecules participating in interspecies quorum sensing was pursued as a next-generation antibiofouling strategy in an MBR for wastewater treatment. We isolated an indigenous QQ bacterium ( Acinetobacter sp. DKY-1) that can attenuate the expression of the quorum-sensing (QS) response through the inactivation of an autoinducer-2 signaling molecule, 4,5-dihydroxy-2,3-pentanedione (DPD), among four kinds of autoinducer-2 QS bacteria. DKY-1 released AI-2 QQ compounds, which were verified to be hydrophilic with a molecular weight of <400 Da. The addition of DKY-1 entrapping beads into an MBR significantly decreased DPD concentration and remarkably reduced membrane biofouling. This new approach, combining molecular biology with wastewater engineering, could enlarge the range of QQ-MBR for antibiofouling and energy savings in the field of wastewater treatment.


Subject(s)
Acinetobacter , Biofouling , Bacteria , Bioreactors , Quorum Sensing , Wastewater
10.
JCI Insight ; 2(23)2017 12 07.
Article in English | MEDLINE | ID: mdl-29212948

ABSTRACT

ER stress has emerged as a signaling platform underlying the pathogenesis of various kidney diseases. Thus, there is an urgent need to develop ER stress biomarkers in the incipient stages of ER stress-mediated kidney disease, when a kidney biopsy is not yet clinically indicated, for early therapeutic intervention. Cysteine-rich with EGF-like domains 2 (CRELD2) is a newly identified protein that is induced and secreted under ER stress. For the first time to our knowledge, we demonstrate that CRELD2 can serve as a sensitive urinary biomarker for detecting ER stress in podocytes or renal tubular cells in murine models of podocyte ER stress-induced nephrotic syndrome and tunicamycin- or ischemia-reperfusion-induced acute kidney injury (AKI), respectively. Most importantly, urinary CRELD2 elevation occurs in patients with autosomal dominant tubulointerstitial kidney disease caused by UMOD mutations, a prototypical tubular ER stress disease. In addition, in pediatric patients undergoing cardiac surgery, detectable urine levels of CRELD2 within postoperative 6 hours strongly associate with severe AKI after surgery. In conclusion, our study has identified CRELD2 as a potentially novel urinary ER stress biomarker with potential utility in early diagnosis, risk stratification, treatment response monitoring, and directing of ER-targeted therapies in selected patient subgroups in the emerging era of precision nephrology.


Subject(s)
Acute Kidney Injury/urine , Cell Adhesion Molecules/urine , Endoplasmic Reticulum Stress/physiology , Extracellular Matrix Proteins/urine , Nephrotic Syndrome/urine , Acute Kidney Injury/diagnosis , Acute Kidney Injury/etiology , Acute Kidney Injury/physiopathology , Animals , Biomarkers/urine , Cardiac Surgical Procedures , Cell Adhesion Molecules/physiology , Child , Extracellular Matrix Proteins/physiology , Humans , Male , Mice, Inbred C57BL , Mutation , Nephritis, Interstitial/genetics , Nephritis, Interstitial/physiopathology , Nephritis, Interstitial/urine , Nephrotic Syndrome/diagnosis , Nephrotic Syndrome/physiopathology , Podocytes/metabolism , Postoperative Complications/urine , Uromodulin/genetics
11.
Transl Res ; 188: 1-9, 2017 10.
Article in English | MEDLINE | ID: mdl-28719799

ABSTRACT

Mesencephalic astrocyte-derived neurotrophic factor (MANF), a newly identified 18-kDa soluble protein, localizes to the luminal endoplasmic reticulum (ER), whose stress can stimulate MANF expression and secretion. In Drosophila and zebrafish, MANF regulates dopaminergic neuron development. In contrast, in mice, MANF deficiency leads to diabetes and activation of the unfolded protein response. Recent studies in rodent models have demonstrated that MANF mitigates diabetes, exerts neurotrophic function in neurodegenerative disease, protects cardiomyocytes and neurons in myocardial infarction and cerebral ischemia, respectively, and promotes immune cell phenotype switch from proinflammatory macrophages to prorepair anti-inflammatory macrophages. The cytoprotective mechanisms of MANF on ER stress are currently under active investigation. In addition, for the first time, we have discovered that MANF can potentially serve as a urinary ER stress biomarker in ER stress-mediated kidney disease. These studies have underscored the diagnostic and therapeutic importance of MANF in ER diseases.


Subject(s)
Endoplasmic Reticulum/metabolism , Nerve Growth Factors/metabolism , Animals , Biomarkers , Humans , Nerve Growth Factors/genetics , Protein Conformation , Species Specificity
12.
Environ Sci Technol ; 50(20): 10914-10922, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27634354

ABSTRACT

In the last 30 years, the use of membrane bioreactors (MBRs) for advanced wastewater treatment and reuse have been expanded continuously, but they still suffer from excessive energy consumption resulting from the intrinsic problem of membrane biofouling. One of the major causes of biofouling in MBRs is bacterial quorum sensing (QS) via N-acylhomoserine lactones (AHLs) and/or autoinducer-2 (AI-2), enabling intra- and interspecies communications, respectively. In this study, we demonstrate that farnesol can substantially mitigate membrane biofouling in a MBR due to its quorum quenching (QQ) activity. When Candida albicans (a farnesol producing fungus) entrapping polymer beads (AEBs) were placed in the MBR, the rate of transmembrane pressure (TMP) rise-up was substantially decreased, even for lower aeration intensities. This finding corresponds to a specific aeration energy savings of approximately 40% (25% through the physical washing effect and a further 15% through the biological QQ effect of AEBs) compared to conventional MBRs without AEBs. A real-time RT-qPCR analysis revealed that farnesol secreted from C. albicans mitigated the biofilm formation in MBRs via the suppression of AI-2 QS. Successful control of biofouling and energy savings through fungal-to-bacterial QQ could be expanded to the plant scale for MBRs in wastewater treatment with economic feasibility.


Subject(s)
Quorum Sensing/drug effects , Wastewater , Biofouling , Bioreactors/microbiology , Membranes, Artificial , Waste Disposal, Fluid
13.
PLoS One ; 11(7): e0158908, 2016.
Article in English | MEDLINE | ID: mdl-27454431

ABSTRACT

Emerging evidence has highlighted the pivotal role of microvasculature injury in the development and progression of renal fibrosis. Angiopoietin-1 (Ang-1) is a secreted vascular growth factor that binds to the endothelial-specific Tie2 receptor. Ang-1/Tie2 signaling is critical for regulating blood vessel development and modulating vascular response after injury, but is dispensable in mature, quiescent vessels. Although dysregulation of vascular endothelial growth factor (VEGF) signaling has been well studied in renal pathologies, much less is known about the role of the Ang-1/Tie2 pathway in renal interstitial fibrosis. Previous studies have shown contradicting effects of overexpressing Ang-1 systemically on renal tubulointerstitial fibrosis when different engineered forms of Ang-1 are used. Here, we investigated the impact of site-directed expression of native Ang-1 on the renal fibrogenic process and peritubular capillary network by exploiting a conditional transgenic mouse system [Pax8-rtTA/(TetO)7 Ang-1] that allows increased tubular Ang-1 production in adult mice. Using a murine unilateral ureteral obstruction (UUO) fibrosis model, we demonstrate that targeted Ang-1 overexpression attenuates myofibroblast activation and interstitial collagen I accumulation, inhibits the upregulation of transforming growth factor ß1 and subsequent phosphorylation of Smad 2/3, dampens renal inflammation, and stimulates the growth of peritubular capillaries in the obstructed kidney. Our results suggest that Ang-1 is a potential therapeutic agent for targeting microvasculature injury in renal fibrosis without compromising the physiologically normal vasculature in humans.


Subject(s)
Angiopoietin-1/genetics , Gene Expression , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Actins/genetics , Actins/metabolism , Animals , Biomarkers , Collagen Type I/genetics , Collagen Type I/metabolism , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Kidney Diseases/metabolism , Mice , Mice, Transgenic , Microcirculation , Neovascularization, Pathologic/genetics , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
14.
J Am Soc Nephrol ; 27(10): 2974-2982, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26940092

ABSTRACT

Endoplasmic reticulum (ER) stress and disrupted proteostasis contribute to the pathogenesis of a variety of glomerular and tubular diseases. Thus, it is imperative to develop noninvasive biomarkers for detecting ER stress in podocytes or tubular cells in the incipient stage of disease, when a kidney biopsy is not yet clinically indicated. Mesencephalic astrocyte-derived neurotrophic factor (MANF) localizes to the ER lumen and is secreted in response to ER stress in several cell types. Here, using mouse models of human nephrotic syndrome caused by mutant laminin ß2 protein-induced podocyte ER stress and AKI triggered by tunicamycin- or ischemia-reperfusion-induced tubular ER stress, we examined MANF as a potential urine biomarker for detecting ER stress in podocytes or renal tubular cells. ER stress upregulated MANF expression in podocytes and tubular cells. Notably, urinary MANF excretion concurrent with podocyte or tubular cell ER stress preceded clinical or histologic manifestations of the corresponding disease. Thus, MANF can potentially serve as a urine diagnostic or prognostic biomarker in ER stress-related kidney diseases to help stratify disease risk, predict disease progression, monitor treatment response, and identify subgroups of patients who can be treated with ER stress modulators in a highly targeted manner.


Subject(s)
Endoplasmic Reticulum Stress , Kidney Diseases/urine , Nerve Growth Factors/urine , Animals , Biomarkers/urine , Kidney Diseases/etiology , Mice
15.
Mol Endocrinol ; 28(6): 822-33, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24725081

ABSTRACT

Thyroid hormone signaling has long been implicated in mammalian testicular function, affecting steroidogenesis in testicular Leydig cells. However, its molecular mechanism is not well understood. Here, we investigated the molecular action of thyroid hormone receptor-α (TRα) on mouse testicular steroidogenesis. TRα/thyroid hormone (T3) signaling differentially affected the expression of steroidogenic enzyme genes, mainly regulating their promoter activity. TRα directly regulated the promoter activity of the cytochrome P450 17α-hydroxylase/C17-20 lyase gene, elevating its expression in the presence of T3. TRα also indirectly regulated the expression of steroidogenic enzyme genes, such as steroidogenic acute regulatory protein and 3ß-hydroxysteroid dehydrogenase, by modulating the transactivation of Nur77 on steroidogenic enzyme gene promoters through protein-protein interaction. TRα enhanced Nur77 transactivation by excluding histone deacetylases from Nur77 in the absence of T3, whereas liganded TRα inhibited Nur77 transactivation, likely due to interfering with the recruitment of coactivator such as the steroid receptor coactivator-1 to Nur77. Together, these findings suggest a role of TRα/T3 in testicular steroidogenesis and may provide molecular mechanisms for the differential regulation of steroidogenic enzyme genes by thyroid hormone.


Subject(s)
Gene Expression Regulation, Enzymologic , Leydig Cells/enzymology , Steroid 17-alpha-Hydroxylase/genetics , Thyroid Hormone Receptors alpha/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP/physiology , HEK293 Cells , Histone Deacetylases/metabolism , Humans , Male , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Protein Binding , Response Elements , Signal Transduction , Steroid 17-alpha-Hydroxylase/metabolism , Transcriptional Activation , Triiodothyronine/physiology
16.
J Am Soc Nephrol ; 24(7): 1127-38, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23661808

ABSTRACT

DLG1 (discs-large homolog 1) and CASK (calcium/calmodulin-dependent serine protein kinase) interact at membrane-cytoskeleton interfaces and function as scaffolding proteins that link signaling molecules, receptors, and other scaffolding proteins at intercellular and synaptic junctions. Dlg1-null mice exhibit hydronephrosis, hydroureter, and occasionally hypoplastic kidneys, whereas Cask-null mice do not. To investigate whether DLG1 and CASK cooperate in the developing urogenital system, we generated mice deficient in both DLG1 and CASK either 1) globally, 2) in metanephric mesenchyme, or 3) in nephron progenitors. With each approach, Dlg1;Cask double-knockout (DKO) kidneys were severely hypoplastic and dysplastic and demonstrated rapid, premature depletion of nephron progenitors/stem cells. Several cellular and molecular defects were observed in the DKO kidneys, including reduced proliferation and increased apoptosis of cells in the nephrogenic zone and a progressive decrease in the number of cells expressing SIX2, a transcription factor essential for maintaining nephron progenitors. Fgf8 expression was reduced in early-stage DKO metanephric mesenchyme, accompanied by reduced levels of components of the Ras pathway, which is activated by fibroblast growth factor (FGF) signaling. Moreover, Dlg1(+/-);Cask(-/-) (het/null) kidneys were moderately hypoplastic and demonstrated impaired aggregation of SIX2-positive cells around the ureteric bud tips. Nephron progenitor-specific het/null mice survived with small kidneys but developed glomerulocystic kidney disease and renal failure. Taken together, these results suggest that DLG1 and CASK play critical cooperative roles in maintaining the nephron progenitor population, potentially via a mechanism involving effects on FGF signaling.


Subject(s)
Cell Differentiation , Guanylate Kinases/metabolism , Nephrons/embryology , Nerve Tissue Proteins/metabolism , Organogenesis , Stem Cells/cytology , Animals , Discs Large Homolog 1 Protein , Gene Expression , Mice , Mice, Knockout , Nephrons/abnormalities , Nephrons/metabolism , SAP90-PSD95 Associated Proteins , Signal Transduction
17.
Toxicology ; 288(1-3): 18-26, 2011 Oct 09.
Article in English | MEDLINE | ID: mdl-21726595

ABSTRACT

Endocrine disruptors (EDs) affect the function of animal reproductive systems. Recently, 2,2',4,4'-tetrahydroxybenzophenone (BP2), which is a component of UV protection products, was found to be an ED that interferes with the thyroid hormone (TH) axis. However, BP2 activity in the testis has not been well addressed. In this study, we have examined the effects of BP2 on steroidogenesis in testicular Leydig cells in connection with thyroid hormone signaling, which is known to play an important role in testicular development and function. Our study showed that BP2 affected the expression of steroidogenic enzyme genes in testicular Leydig cells, which is differentially regulated by thyroid hormone/thyroid hormone receptor (TR) signaling. In MA-10 Leydig cell line, TR/T3 signaling increased the expression of P450c17 and P450scc, while it decreased the expression of StAR and 3ß-HSD. Interestingly, BP2 affected the expression of steroidogenic enzyme genes in a manner opposite to that of T3 signaling. BP2 downregulated the TRα/T3-activation of P450c17 and P450scc expression while enhancing the TRα/T3-repression of StAR and 3ß-HSD expression. Transient transfection analyses with promoter-reporter constructs revealed that BP2 altered the expression of steroidogenic enzyme genes by affecting the cAMP and Nur77-activated promoter activity of P450c17, StAR, and 3ß-HSD. Animal experiments with mice revealed that BP2 decreased the production of testosterone in the testis by affecting the expression of some steroidogenic enzyme genes in vivo. Together, these findings elucidate a molecular mechanism of BP2 action underlying testicular steroidogenesis and also suggest that BP2 acts, in part, as a thyroid antagonist that affects steroidogenesis in the testis.


Subject(s)
Benzophenones/toxicity , Endocrine Disruptors/toxicity , Leydig Cells/drug effects , Testis/drug effects , Testosterone/biosynthesis , Animals , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Thyroid Hormone/drug effects , Receptors, Thyroid Hormone/metabolism , Steroids/biosynthesis , Testis/metabolism , Transfection , Triiodothyronine/drug effects , Triiodothyronine/metabolism
18.
J Biol Chem ; 285(29): 22360-9, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20472563

ABSTRACT

ARR19 (androgen receptor corepressor-19 kDa), a leucine-rich protein whose expression is down-regulated by luteinizing hormone and cAMP, is differentially expressed during the development of Leydig cells and inhibits testicular steroidogenesis by reducing the expression of steroidogenic enzymes. However, the molecular events behind the suppression of testicular steroidogenesis are unknown. In the present study, we demonstrate that ARR19 inhibits the transactivation of orphan nuclear receptor Nur77, which is one of the major transcription factors that regulate the expression of steroidogenic enzyme genes in Leydig cells. ARR19 physically interacts with Nur77 and suppresses Nur77-induced promoter activity of steroidogenic enzyme genes including StAR, P450c17, and 3beta-HSD in Leydig cells. Transient transfection and chromatin immunoprecipitation assays revealed that ARR19-mediated reduced expression of steroidogenic enzyme genes was likely due to the interference of SRC-1 recruitment to Nur77 protein on the promoter of steroidogenic enzyme genes. These findings suggest that ARR19 acts as a novel coregulator of Nur77, in turn regulating Nur77-induced testicular steroidogenesis, and may play an important role in the development and function of testicular Leydig cells.


Subject(s)
Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Repressor Proteins/metabolism , Steroids/biosynthesis , Testis/metabolism , Transcriptional Activation/genetics , Adenoviridae/metabolism , Animals , Binding, Competitive , Cell Nucleus/metabolism , Leydig Cells/cytology , Leydig Cells/metabolism , MARVEL Domain-Containing Proteins , Male , Membrane Proteins , Mice , Nuclear Receptor Coactivator 1/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport , Repressor Proteins/chemistry , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Testis/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...